Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Chem Neuroanat ; 129: 102237, 2023 04.
Article in English | MEDLINE | ID: mdl-36736441

ABSTRACT

The number of people diagnosed with metabolic syndrome (MetS) has increased dramatically to reach alarming proportions worldwide. The origin of MetS derives from bad eating habits and sedentary lifestyle. Most people consume foods high in carbohydrates and saturated fat. In recent years, it has been reported that alterations in insulin at the brain level could have an impact on the appearance of neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, dementia, depression, and other types of disorders that compromise brain function. These alterations have been associated with damage to the structure and function of neurons located in the reptilian and limbic systems, a decrease in dendritic arborization and an exacerbated inflammatory state that impaired learning and memory and increased in the state of stress and anxiety. Although the molecular mechanisms induced by MetS to cause neurodegeneration are not fully understood. The aim of this study is to know the effect of the intake of hypercaloric diets on the structure and function of neurons located in the frontal cortex, hypothalamus and hippocampus and its impact on behavior in rats with metabolic syndrome. In conclusion, the present study illustrated that chronic exposure to hypercaloric diets, with a high content of sugars and saturated fatty acids, induces a proinflammatory state and exacerbates oxidative stress in brain regions such as the hypothalamus, hippocampus, and frontal cortex, leading to dysfunction. metabolism, neuronal damage, and recognition memory loss.


Subject(s)
Alzheimer Disease , Metabolic Syndrome , Animals , Rats , Carbohydrates , Diet , Diet, High-Fat , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , Neurons/metabolism , Memory Disorders/metabolism
2.
Neurochem Res ; 46(5): 1151-1165, 2021 May.
Article in English | MEDLINE | ID: mdl-33559829

ABSTRACT

The consumption of foods rich in carbohydrates, saturated fat, and sodium, accompanied by a sedentary routine, are factors that contribute to the progress of metabolic syndrome (MS). In this way, they cause the accumulation of body fat, hypertension, dyslipidemia, and hyperglycemia. Additionally, MS has been shown to cause oxidative stress, inflammation, and death of neurons in the hippocampus. Consequently, spatial and recognition memory is affected. It has recently been proposed that metformin decavanadate (MetfDeca) exerts insulin mimetic effects that enhance metabolism in MS animals; however, what effects it can cause on the hippocampal neurons of rats with MS are unknown. The objective of the work was to evaluate the effect of MetfDeca on hippocampal neurodegeneration and recognition memory in rats with MS. Administration of MetfDeca for 60 days in MS rats improved object recognition memory (NORt). In addition, MetfDeca reduced markers of oxidative stress and hippocampal neuroinflammation. Accompanied by an increase in the density and length of the dendritic spines of the hippocampus of rats with MS. We conclude that MetfDeca represents an important therapeutic agent to treat MS and induce neuronal and cognitive restoration mechanisms.


Subject(s)
Memory/drug effects , Metabolic Syndrome/drug therapy , Metformin/therapeutic use , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Vanadates/therapeutic use , Animals , Catalase/metabolism , Drug Combinations , Hippocampus/drug effects , Inflammation/drug therapy , Inflammation/etiology , Inflammation/pathology , Male , Metabolic Syndrome/complications , Metabolic Syndrome/pathology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , Rats, Wistar , Superoxide Dismutase/drug effects
3.
Synapse ; 75(2): e22186, 2020 02.
Article in English | MEDLINE | ID: mdl-32780904

ABSTRACT

Metabolic syndrome (MS) results from excessive consumption of high-calorie foods and sedentary lifestyles. Clinically, insulin resistance, abdominal obesity, hyperglycemia, dyslipidemia, and hypertension are observed. MS has been considered a risk factor in the development of dementia. In the brain, a metabolically impaired environment generates oxidative stress and excessive production of pro-inflammatory cytokines that deteriorate the morphology and neuronal function in the hippocampus, leading to cognitive impairment. Therapeutic alternatives suggest that phenolic compounds can be part of the treatment for neuropathies and metabolic diseases. In recent years, the use of Gallic Acid (GA) has demonstrated antioxidant and anti-inflammatory effects that contribute to neuroprotection and memory improvement in animal models. However, the effect of GA on hippocampal neurodegeneration and memory impairment under MS conditions is still unclear. In this work, we administered GA (20 mg/kg) for 60 days to rats with MS. The results show that GA treatment improved zoometric and biochemical parameters, as well as the recognition memory, in animals with MS. Additionally, GA administration increased hippocampal dendritic spines and decreased oxidative stress and inflammation. Our results show that GA treatment improves metabolism: reducing the oxidative and inflammatory environment that facilitates the recovery of the neuronal morphology in the hippocampus of rats with MS. Consequently, the recognition of objects by these animals, suggesting that GA could be used therapeutically in metabolic disorders that cause dementia.


Subject(s)
Gallic Acid/pharmacology , Hippocampus/drug effects , Metabolic Syndrome/metabolism , Recognition, Psychology/drug effects , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Catalase/drug effects , Catalase/metabolism , Dendrites/drug effects , Dendrites/pathology , Hippocampus/metabolism , Hippocampus/pathology , Inflammation/metabolism , Insulin/blood , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Memory/drug effects , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/metabolism , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
4.
Synapse ; 74(9): e22153, 2020 09.
Article in English | MEDLINE | ID: mdl-32190918

ABSTRACT

Metabolic syndrome (MS) is a health problem that is characterized by body fat accumulation, hypertension, dyslipidemia, and hyperglycemia; recently, it has been demonstrated that MS also damages memory processes. The first-line drug in the treatment of MS and type 2 diabetes mellitus is metformin, which is an antihyperglycemic agent. This drug has been shown to produce neuroprotection and to improve memory processes. However, the mechanism involved in this neuroprotection is unknown. A 90-day administration of metformin improved the cognitive processes of rats with MS as evaluated by the novel object recognition test, and this finding could be explained by an increase in the neuronal spine density and spine length. We also found that metformin increased the immunoreactivity of synaptophysin, sirtuin-1, AMP-activated protein kinase, and brain-derived neuronal factor, which are important plasticity markers. We conclude that metformin is an important therapeutic agent that increases neural plasticity and protects cognitive processes. The use of this drug is important in the minimization of the damage caused by MS.


Subject(s)
Hippocampus/drug effects , Hypoglycemic Agents/pharmacology , Metabolic Syndrome/physiopathology , Metformin/pharmacology , Neuronal Plasticity , Neuroprotective Agents/pharmacology , Recognition, Psychology , AMP-Activated Protein Kinase Kinases , Animals , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/metabolism , Hippocampus/physiology , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/therapeutic use , Male , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism , Metformin/administration & dosage , Metformin/therapeutic use , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Protein Kinases/metabolism , Rats , Rats, Wistar , Sirtuin 1/metabolism , Synaptophysin/metabolism
5.
Oxid Med Cell Longev ; 2018: 1358057, 2018.
Article in English | MEDLINE | ID: mdl-30154946

ABSTRACT

An important worldwide health problem as the result of current lifestyle is metabolic syndrome (MS). It has been shown that MS induced by a high-calorie diet (HCD) in rats produces cognitive deterioration in the novel object recognition test (NORt) and decreases synaptic connections and dendritic order in the hippocampus and temporal cortex. However, it is unknown whether MS induced by an HCD participates in the cognitive process observed with the injection of Aß1-42 into the hippocampus of rats as a model of Alzheimer disease (AD). The induction of MS in rats produces a deterioration in NORt; however, rats with MS injected with Aß1-42 show a major deterioration in the cognitive process. This event could be explained by the increment in the oxidative stress in both cases studied (MS and Aß1-42): together, the hippocampus and temporal cortex produce an enhancer effect. In the same way, we observed an increment in interleukin-1ß, TNF-α, and GFAP, indicative of exacerbated inflammatory processes by the combination of MS and Aß1-42. We can conclude that MS might play a key role in the apparition and development of cognitive disorders, including AD. We propose that metabolic theory is important to explain the apparition of cognitive diseases.


Subject(s)
Amyloid beta-Peptides/therapeutic use , Inflammation/pathology , Memory Disorders/etiology , Metabolic Syndrome/complications , Oxidative Stress/physiology , Amyloid beta-Peptides/pharmacology , Animals , Disease Models, Animal , Hippocampus/drug effects , Male , Memory Disorders/pathology , Rats , Rats, Wistar
6.
Synapse ; 71(10): e21990, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28650104

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia and one of the most important causes of morbidity and mortality among the aging population. AD diagnosis is made post-mortem, and the two pathologic hallmarks, particularly evident in the end stages of the illness, are amyloid plaques and neurofibrillary tangles. Currently, there is no curative treatment for AD. Additionally, there is a strong relation between oxidative stress, metabolic syndrome, and AD. The high levels of circulating lipids and glucose imbalances amplify lipid peroxidation that gradually diminishes the antioxidant systems, causing high levels of oxidative metabolism that affects cell structure, leading to neuronal damage. Accumulating evidence suggests that AD is closely related to a dysfunction of both insulin signaling and glucose metabolism in the brain, leading to an insulin-resistant brain state. Four drugs are currently used for this pathology: Three FDA-approved cholinesterase inhibitors and one NMDA receptor antagonist. However, wide varieties of antioxidants are promissory to delay or prevent the symptoms of AD and may help in treating the disease. Therefore, therapeutic efforts to achieve attenuation of oxidative stress could be beneficial in AD treatment, attenuating Aß-induced neurotoxicity and improve neurological outcomes in AD. The term inflammaging characterizes a widely accepted paradigm that aging is accompanied by a low-grade chronic up-regulation of certain pro-inflammatory responses in the absence of overt infection, and is a highly significant risk factor for both morbidity and mortality in the elderly.

7.
Oxid Med Cell Longev ; 2016: 8725354, 2016.
Article in English | MEDLINE | ID: mdl-27069534

ABSTRACT

Energy drinks (EDs) are often consumed in combination with alcohol because they reduce the depressant effects of alcohol. However, different researches suggest that chronic use of these psychoactive substances in combination with alcohol can trigger an oxidative and inflammatory response. These processes are regulated by both a reactive astrogliosis and an increase of proinflammatory cytokines such as IL-1ß, TNF-α, and iNOS, causing cell death (apoptosis) at the central and peripheral nervous systems. Currently, mechanisms of toxicity caused by mixing alcohol and ED in the brain are not well known. In this study, we evaluated the effect of chronic alcohol consumption in combination with ED on inflammatory response and oxidative stress in the temporal cortex (TCx) and hippocampus (Hp) of adult rats (90 days old). Our results demonstrated that consuming a mixture of alcohol and ED for 60 days induced an increase in reactive gliosis, IL-1ß, TNF-α, iNOS, reactive oxygen species, lipid peroxidation, and nitric oxide, in the TCx and Hp. We also found immunoreactivity to caspase-3 and a decrease of synaptophysin in the same brain regions. The results suggested that chronic consumption of alcohol in combination with ED causes an inflammatory response and oxidative stress, which induced cell death via apoptosis in the TCx and Hp of the adult rats.


Subject(s)
Energy Drinks/adverse effects , Ethanol/adverse effects , Hippocampus/pathology , Inflammation/pathology , Oxidative Stress , Temporal Lobe/pathology , Animals , Caspase 3/metabolism , Cytokines/metabolism , Ethanol/blood , Glial Fibrillary Acidic Protein/metabolism , Inflammation Mediators/metabolism , Male , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Rats, Wistar , Synaptophysin/metabolism
8.
Synapse ; 69(9): 421-33, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26073877

ABSTRACT

A high calorie intake can induce the appearance of the metabolic syndrome (MS), which is a serious public health problem because it affects glucose levels and triglycerides in the blood. Recently, it has been suggested that MS can cause complications in the brain, since chronic hyperglycemia and insulin resistance are risk factors for triggering neuronal death by inducing a state of oxidative stress and inflammatory response that affect cognitive processes. This process, however, is not clear. In this study, we evaluated the effect of the consumption of a high-calorie diet (HCD) on both neurodegeneration and spatial memory impairment in rats. Our results demonstrated that HCD (90 day consumption) induces an alteration of the main energy metabolism markers, indicating the development of MS in rats. Moreover, an impairment of spatial memory was observed. Subsequently, the brains of these animals showed activation of an inflammatory response (increase in reactive astrocytes and interleukin1-ß as well as tumor necrosis factor-α) and oxidative stress (reactive oxygen species and lipid peroxidation), causing a reduction in the number of neurons in the temporal cortex and hippocampus. Altogether, these results suggest that a HCD promotes the development of MS and contributes to the development of a neurodegenerative process and cognitive failure. In this regard, it is important to understand the relationship between MS and neuronal damage in order to prevent the onset of neurodegenerative disorders.


Subject(s)
Diet/adverse effects , Hippocampus/metabolism , Memory Disorders/metabolism , Metabolic Diseases/metabolism , Oxidative Stress/physiology , Temporal Lobe/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/pathology , Interleukin-1beta/metabolism , Lipid Peroxidation/physiology , Male , Memory Disorders/etiology , Memory Disorders/pathology , Metabolic Diseases/etiology , Metabolic Diseases/pathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neuroimmunomodulation/physiology , Rats, Wistar , Reactive Oxygen Species/metabolism , Spatial Learning/physiology , Spatial Memory/physiology , Temporal Lobe/pathology , Tumor Necrosis Factor-alpha/metabolism
9.
Synapse ; 69(3): 103-14, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25522178

ABSTRACT

The cannabinoid CB1 (CB1R) and dopaminergic D2 (D2R) receptors modify GABAergic transmission in the globus pallidus. Although dopaminergic denervation produces changes in the expression and supersensitization of these receptors, the consequences of these changes on GABAergic neurotransmission are unknown. The aim of this study was to show the effects of CB1R and D2R activation and coactivation on the uptake and release of [(3) H]GABA in the globus pallidus of hemiparkinsonian rats as well as their effects on motor behavior. The activation of CB1R blocked GABA uptake and decreased GABA release in the globus pallidus in the dopamine denervated side, whereas the co-activation of CB1R-D2R increased GABA release and had no effect on GABA uptake. A microinjection of the CB1R agonist ACEA into the globus pallidus ipsilaterally to a 6-OHDA lesion potentiated turning behavior that was induced by methamphetamine. However, a microinjection of the D2R agonist quinpirole did not modify this behavior, and a microinjection of a mixture of CB1R and D2R agonists significantly potentiated turning behavior. The behavioral effects produced after the activation of the CB1R and the co-activation of CB1R and D2R can be explained by increased GABAergic neurotransmission produced by a block of GABA uptake and an increase in the release of GABA in the globus pallidus, respectively.


Subject(s)
GABAergic Neurons/metabolism , Globus Pallidus/metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptors, Dopamine D2/metabolism , Synaptic Transmission , Animals , Arachidonic Acids/pharmacology , GABAergic Neurons/drug effects , Globus Pallidus/drug effects , Globus Pallidus/physiology , Male , Methamphetamine/pharmacology , Movement , Oxidopamine/toxicity , Quinpirole/pharmacology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/agonists , Receptors, Dopamine D2/agonists , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...